SMYD1 belongs to the SMYD family of methyltransferases, characterized by a split SET domain interrupted by a MYND zinc-finger domain. This unique architecture enables its dual role as a histone methyltransferase (H3K4me) and a transcriptional regulator. SMYD1 interacts with substrates like skNAC (skeletal muscle-specific NAC) and Isl1 (ISL LIM Homeobox 1), influencing gene expression critical for muscle differentiation and heart development .
These antibodies are pivotal in studying SMYD1’s localization, interactions, and functional roles. Key applications include:
skNAC Methylation: SMYD1 methylates skNAC at lysine 1975, enhancing transcription of myoglobin (Mb), a muscle-specific oxygen transporter. Antibodies confirmed SMYD1-skNAC interaction in C2C12 myoblasts .
Myosin Heavy-Chain (MHC) Regulation: SMYD1b (a SMYD1 isoform) is essential for MHC protein stability in craniofacial muscles. Antibody-based studies revealed reduced MHC expression in smyd1b mutants .
LPS-Induced IL-6 Expression: SMYD1 upregulates interleukin-6 (IL-6) in endothelial cells (ECs) via HDAC-mediated repression. Antibodies demonstrated SMYD1’s nuclear translocation in LPS-stimulated ECs .
Angiogenesis: SMYD1 interacts with SRF (serum response factor) in ECs to promote tube formation. Antibodies localized SMYD1 to nuclear regions critical for vascular remodeling .
Isl1 Promoter Binding: SMYD1 binds the Isl1 promoter via its MYND domain, activating transcription. ChIP-qPCR with SMYD1 antibodies identified enrichment at conserved binding sites (e.g., SB4) .
ANF Repression: SMYD1 represses ANF (atrial natriuretic factor) expression in cardiomyocytes. Antibody assays linked this repression to HDAC recruitment .
SMYD1b vs. SMYD1a: SMYD1b, but not SMYD1a, methylates skNAC and regulates Mb expression. Antibody-based assays distinguished isoform-specific activities in myoblast differentiation .
Domain Requirements: The SET (catalytic) and MYND (binding) domains are essential for skNAC methylation. Mutagenesis studies with SMYD1 antibodies confirmed domain-specific roles .
Cardiomyopathies: SMYD1 dysfunction disrupts sarcomere assembly and myosin expression, leading to structural heart defects. Antibodies have been used to study SMYD1’s role in zebrafish and mouse models .
Inflammatory Pathways: SMYD1’s involvement in LPS-triggered cytokine release suggests therapeutic potential in vascular inflammation .
SMYD1 (SET and MYND domain-containing protein 1) is a histone-lysine N-methyltransferase that functions as a transcriptional repressor. It is essential for cardiomyocyte differentiation and cardiac morphogenesis, playing a key role in myogenesis - the process by which muscle cells differentiate and mature. Its critical involvement in muscle formation and maintenance makes it a valuable target for studies on muscular disorders, cardiac diseases, and muscular dystrophies . When designing experiments targeting SMYD1, researchers should consider its dual localization in both cytoplasm and nucleus, as well as its multiple biological functions in chromatin remodeling, transcription regulation, and heart development.
SMYD1 is known by several synonyms in scientific literature and databases:
BOP (CD8 beta opposite)
KMT3D
ZMYND18 (zinc finger, MYND domain containing 18)
ZMYND22
Important identifier information includes:
UniProt Code: Q8NB12
NCBI Gene ID: 150572
NCBI Accession: Q8NB12.1
Chromosomal Location: 2p11.2
Calculated Molecular Weight: 56kDa
Understanding these alternative nomenclatures is essential when conducting literature searches or database queries to ensure comprehensive coverage of SMYD1-related research.
SMYD1 exhibits multiple biological functions that make it important in developmental and physiological processes:
Transcriptional repression: Acts as a transcriptional repressor in multiple contexts
Histone methylation: Functions as a histone-lysine N-methyltransferase (EC 2.1.1.43)
Cardiac development: Essential for cardiomyocyte differentiation and cardiac morphogenesis
Muscle development: Critical regulator of myoblast differentiation (positive regulation)
Inflammatory response: Contributes to LPS-triggered expression and secretion of IL-6 in endothelial cells
Epigenetic regulation: Affects the H3K4me3 methylation pattern of the IL-6 promoter
When designing experiments to study SMYD1 function, researchers should consider which specific activity they are targeting and select appropriate readouts accordingly.
SMYD1 plays a significant role in inflammatory responses in endothelial cells through several mechanisms:
LPS stimulation upregulates SMYD1 expression in endothelial cells, as demonstrated in EA.hy926 cells
SMYD1 contributes to LPS-triggered expression and secretion of IL-6
SMYD1 induces IL-6 expression through both NF-κB-dependent and NF-κB-independent pathways
The methyltransferase activity of SMYD1 is directly involved in regulating IL-6 expression
SMYD1 affects the H3K4me3 (histone H3 lysine 4 trimethylation) pattern at the IL-6 promoter
For researchers investigating this pathway, experimental designs should include methyltransferase activity assays, chromatin immunoprecipitation (ChIP) for H3K4me3 at the IL-6 promoter, and analysis of NF-κB activation status alongside SMYD1 expression manipulation.
SMYD1's histone-lysine N-methyltransferase activity is central to many of its biological functions. Research has demonstrated:
The methyltransferase domain is essential for SMYD1's role in regulating gene expression
SMYD1 methyltransferase activity directly influences the H3K4me3 methylation pattern at specific gene promoters, including IL-6
Experiments with methyltransferase-deficient SMYD1 mutants (HMTase mutants) show altered regulatory capacity on target genes
SMYD1 can affect gene expression through both methyltransferase-dependent and independent mechanisms
When investigating SMYD1 functions, researchers should consider including methyltransferase-dead mutants as controls to distinguish between enzymatic and scaffolding functions of the protein.
SMYD1 exhibits dynamic subcellular localization that can be altered by cellular stimulation:
Under basal conditions, SMYD1 is detected in both cytoplasm and nucleus
Upon LPS stimulation (1 μg/mL for 3h), SMYD1 shows increased nuclear and cytoplasmic expression in endothelial cells
Quantitative analysis reveals that LPS stimulation significantly increases SMYD1 immunoreactivity in both nuclear and cytosolic compartments
This translocation pattern suggests potential distinct functions in different cellular compartments
For proper analysis of SMYD1 localization, researchers should employ both immunocytochemistry and subcellular fractionation followed by immunoblotting to confirm compartment-specific enrichment.
For optimal detection of SMYD1 using immunoblotting techniques:
Sample preparation:
Total cell lysates should be prepared in a buffer containing protease inhibitors
Protein concentration should be determined using Bradford or BCA assay
Load 20-40 μg of total protein per lane
Electrophoresis and transfer:
Use 10% SDS-PAGE for optimal separation
Transfer to PVDF or nitrocellulose membrane at 100V for 1 hour
Antibody incubation:
Block membranes with 5% non-fat dry milk or BSA
Use anti-SMYD1 antibody at a dilution of 1:200 to 1:2000
Incubate overnight at 4°C for primary antibody
Use appropriate HRP-conjugated secondary antibody (e.g., anti-rabbit IgG)
Detection:
This protocol can be adjusted based on specific experimental needs and the particular anti-SMYD1 antibody being used.
For immunohistochemistry (IHC) and immunofluorescence (IF) detection of SMYD1:
Sample preparation:
For tissues: Fix in 4% paraformaldehyde, paraffin-embed or freeze, and section (4-6 μm)
For cells: Grow on coverslips, fix with 4% paraformaldehyde for 15 minutes at room temperature
Antigen retrieval (for IHC):
Heat-induced epitope retrieval in citrate buffer (pH 6.0) or EDTA buffer (pH 9.0)
Microwave for 15-20 minutes or pressure cooker for 3-5 minutes
Blocking and permeabilization:
Permeabilize with 0.1-0.5% Triton X-100 in PBS for 10 minutes
Block with 5-10% normal serum (from the species of secondary antibody) for 1 hour
Antibody incubation:
For IHC: Anti-SMYD1 antibody at 1:20 to 1:200 dilution
For IF: Anti-SMYD1 antibody at 1:20 to 1:100 dilution
Incubate overnight at 4°C
Detection:
For IHC: Use appropriate HRP-conjugated secondary antibody and DAB substrate
For IF: Use fluorescently-labeled secondary antibodies (e.g., FITC-conjugated anti-rabbit IgG)
Counterstain nuclei with DAPI or Draq5
Recommended positive control tissues:
These protocols should be optimized for specific applications and tissue types.
To effectively manipulate SMYD1 expression for functional studies:
Overexpression approaches:
Transfection with pCMV2-Smyd1-flag vector in cell lines
Adenoviral or lentiviral vectors for harder-to-transfect cells
Inducible expression systems (e.g., Tet-On) for temporal control
Knockdown/silencing strategies:
Transfection with SMYD1-specific siRNA (validated for efficacy)
shRNA delivered via lentiviral vectors for stable knockdown
CRISPR-Cas9 system for gene knockout studies
Verification of manipulation:
RT-qPCR to quantify SMYD1 mRNA levels
Immunoblotting to confirm protein level changes
Functional assays to validate consequences of expression changes
Experimental controls:
These approaches have been validated in endothelial cell models and can be adapted for other cell types depending on transfection efficiency.
When encountering inconsistent SMYD1 antibody staining patterns:
Antibody validation:
Confirm antibody specificity using positive and negative control samples
Validate with knockdown/knockout samples to ensure specificity
Consider using multiple antibodies targeting different epitopes
Protocol optimization:
Test multiple fixation methods (paraformaldehyde, methanol, acetone)
Optimize antigen retrieval conditions (pH, duration, temperature)
Titrate antibody concentration to determine optimal dilution
Extend primary antibody incubation time (overnight at 4°C)
Signal enhancement:
Use signal amplification systems (e.g., tyramide signal amplification)
Optimize exposure settings for imaging
Consider using alternative detection systems
Background reduction:
Increase blocking time and concentration
Add 0.1-0.3% Triton X-100 to antibody diluent
Include additional washing steps with higher salt concentration
Subcellular localization considerations:
Documenting all optimization steps will help establish reproducible protocols for future experiments.
To accurately distinguish between SMYD1 isoforms and related SMYD family proteins:
Isoform identification:
Use isoform-specific primers for RT-PCR
Employ antibodies targeting isoform-specific regions
Analyze molecular weight differences on Western blots (56-57 kDa for SMYD1)
Family member discrimination:
Select antibodies validated for lack of cross-reactivity with other SMYD family members
Include positive controls for each SMYD family member
Use siRNA knockdown of specific SMYD proteins as negative controls
Sequence analysis considerations:
SMYD1 shares sequence similarity with other SMYD family members
The SET domain is highly conserved across the family
The MYND domain provides some distinction between family members
Expression pattern analysis:
Combining multiple approaches provides the most reliable discrimination between closely related proteins.
When analyzing SMYD1's role in inflammatory responses, researchers should consider:
Stimulus-specific effects:
LPS concentration-dependent effects (1 ng/mL to 10 μg/mL)
Time-dependent changes (short-term vs. long-term exposure)
Different inflammatory stimuli may yield varying responses
Cell type considerations:
Endothelial cells (such as EA.hy926) show specific responses
Primary vs. immortalized cell differences
Tissue-specific endothelial cells may respond differently
Pathway analysis:
NF-κB-dependent mechanisms
NF-κB-independent mechanisms
Interaction with other inflammatory signaling pathways
Epigenetic analysis:
H3K4me3 modifications at specific promoters
Changes in DNA accessibility
Integration with transcription factor binding
Downstream effects measurement:
A comprehensive analysis should include both molecular mechanisms and functional outcomes to fully characterize SMYD1's role in inflammation.
SMYD1's essential role in cardiac development suggests several promising research directions:
Cardiac regeneration:
SMYD1's potential role in cardiac progenitor cell differentiation
Application in directing stem cell fate toward cardiomyocyte lineage
Development of SMYD1-targeting strategies to enhance cardiac repair
Heart failure models:
Changes in SMYD1 expression and activity in different heart failure models
Correlation with cardiac remodeling and fibrosis
Potential therapeutic targeting to mitigate adverse remodeling
Inflammatory cardiac conditions:
SMYD1's contribution to inflammatory signaling in cardiac tissue
Role in myocarditis and other inflammatory heart diseases
Interaction with cardiac immune cell populations
Developmental cardiac defects:
These research areas may yield new insights into cardiac pathophysiology and identify novel therapeutic strategies.
Integrating multi-omics approaches can provide comprehensive insights into SMYD1 function:
Epigenomic approaches:
ChIP-seq for genome-wide mapping of SMYD1 binding sites
CUT&RUN for higher resolution protein-DNA interaction mapping
ATAC-seq to correlate SMYD1 activity with chromatin accessibility changes
Transcriptomic analysis:
RNA-seq following SMYD1 manipulation to identify regulated gene networks
Single-cell RNA-seq to capture cell-specific responses
Nascent RNA analysis to distinguish direct from indirect effects
Proteomic strategies:
IP-MS to identify SMYD1 protein interaction partners
Phosphoproteomics to map signaling cascades affected by SMYD1
Proteome-wide analysis of histone modifications
Metabolomic considerations:
Analysis of metabolic changes in SMYD1-manipulated systems
Connection between cellular metabolism and SMYD1 activity
SAM (S-adenosyl methionine) availability effects on SMYD1 function
Integrated data analysis:
These approaches can reveal previously unrecognized functions and regulatory mechanisms of SMYD1.
When selecting SMYD1 antibodies for research applications, consider these technical specifications:
Always validate antibodies in your specific experimental system before conducting critical experiments.
For studying SMYD1's methyltransferase activity, the following experimental conditions are recommended:
Enzyme preparation:
Recombinant SMYD1 expression in E. coli or baculovirus systems
FLAG-tagged or His-tagged protein for purification
Active enzyme requires proper folding and possible co-factors
Assay conditions:
Buffer: 50 mM Tris-HCl (pH 8.0), 10% glycerol, 20 mM KCl, 5 mM MgCl₂
Temperature: 30°C is optimal for enzymatic activity
Incubation time: 30-60 minutes for standard assays
Substrates:
S-adenosyl methionine (SAM) as methyl donor
Recombinant histones (especially H3)
Synthetic histone peptides containing target lysine residues
Activity detection methods:
Radioactive assay using ³H-SAM
Antibody-based detection of methylated histones
Mass spectrometry to identify methylation sites
Fluorescence-based assays for high-throughput screening
Controls:
These conditions should be optimized for specific experimental objectives and available resources.
The SMYD protein family shares structural similarities but exhibits distinct functional properties:
| Feature | SMYD1 | SMYD2 | SMYD3 | SMYD4 | SMYD5 |
|---|---|---|---|---|---|
| Primary Methylation Activity | H3K4 | H3K4, H3K36, non-histone targets | H3K4, H4K5 | Not fully characterized | H4K20 |
| Expression Pattern | Primarily muscle, heart | Broadly expressed | Broad with upregulation in cancer | Broad | Broad |
| Developmental Role | Critical for cardiac and skeletal muscle | Cell proliferation | Cardiac and skeletal muscle | Development, tumor suppression | Immune regulation |
| Subcellular Localization | Nucleus and cytoplasm | Nucleus and cytoplasm | Primarily nuclear | Cytoplasmic | Nuclear |
| Role in Transcription | Repressor | Repressor | Activator | Repressor | Repressor |
| Inflammatory Regulation | Promotes IL-6 expression | Limited evidence | Context-dependent | Not well characterized | Immune cell regulation |
| Disease Association | Cardiac disorders | Cancer | Cancer, cardiac hypertrophy | Cancer (tumor suppressor) | Inflammatory disorders |
This comparative analysis highlights the unique role of SMYD1 in muscle development and function, distinguishing it from other family members that have broader expression patterns and functions.
When faced with contradictory results in SMYD1 research across different experimental systems, consider these interpretative frameworks:
Cell type-specific effects:
SMYD1 may have different binding partners in different cell types
Chromatin landscape varies between cell types, affecting SMYD1 target accessibility
Signal transduction pathways may differ, altering SMYD1 regulation
Experimental condition variables:
Culture conditions (serum levels, oxygen tension, confluency)
Stimulus concentrations and duration (e.g., LPS dosage)
Acute vs. chronic manipulation of SMYD1 expression
Methodological considerations:
Antibody specificity issues
Detection method sensitivity
Overexpression artifacts vs. physiological levels
Isoform-specific effects:
Different SMYD1 isoforms may have distinct functions
Expression ratios of isoforms may vary between systems
Post-translational modifications may differ
Integration approach: